1
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
2
|
Li R, Lv W, Wang DL, Chen N. A Systematic Review of Immune Cell Roles in Breast Cancer Immunotherapy. Cancer Rep (Hoboken) 2025; 8:e70217. [PMID: 40356222 PMCID: PMC12069222 DOI: 10.1002/cnr2.70217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent malignancy among women and is associated with high mortality and significant clinical challenges. Although conventional treatments such as surgery, chemotherapy, and radiotherapy have significantly improved patient survival, their efficacy remains limited by severe side effects and treatment resistance. In recent years, advances in immunotherapy have underscored the pivotal role of immune cells in treating BC. RECENT FINDINGS This systematic review summarizes the current knowledge on the roles of immune cells within the BC tumor microenvironment (TME), including their phenotypes, functions, and implications for immunotherapy. Following PRISMA guidelines, 71 studies published between 2010 and 2024 were analyzed. The results indicate that immune cell populations-such as tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), natural killer (NK) cells, dendritic cells (DCs), and myeloid-derived suppressor cells (MDSCs)-are integral to tumor progression and therapeutic response. However, their functional heterogeneity and plasticity remain key obstacles to the development of effective and personalized immunotherapeutic strategies. CONCLUSION Further research is needed to clarify the mechanisms governing immune cell behavior within the BC TME and to advance precision immunotherapy. Such insights will lay the foundation for individualized treatment approaches, ultimately improving patient outcomes and quality of life (QoL).
Collapse
Affiliation(s)
- Rui Li
- Shandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Wei Lv
- Department of General SurgeryShandong Provincial HospitalJinanShandongChina
| | | | - Na Chen
- Department of Internal MedicineShandong Provincial HospitalJinanShandongChina
| |
Collapse
|
3
|
Zhang Z, Li M, Zhang L, Wang M, Liu D, Tang S, Li Y, Fang X, Ren S. Promising future of breast cancer vaccine asking for multidisciplinary collaboration: a literature review. Front Cell Dev Biol 2025; 13:1578883. [PMID: 40342927 PMCID: PMC12058682 DOI: 10.3389/fcell.2025.1578883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
Despite improved efficacy of breast cancer vaccine (BCV) made by multidisciplinary collaboration from fields such as materials science and computer science, clinical translation is still far from satisfactory. Herein, we reviewed the evolution trajectory of BCV and summarized the frontier topics and challenges for achieving successful clinical translation. Our analysis suggests that multi-omics and immunoinformatics are increasingly being used to expand target repertoires, and dedicated vaccine platforms are facilitating precise spatiotemporal co-delivery of epitopes and immune modulators. BCV has evolved towards precise delivery, strong immune properties, and combined therapy. Clinical translation requires the joint efforts of experts in clinical oncology, immunology, pharmacology, materials, and computer science.
Collapse
Affiliation(s)
- Zhanyi Zhang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mengyuan Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Lei Zhang
- State Key Laboratory of Primate Biomedical Research, Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Maohua Wang
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dequan Liu
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shicong Tang
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuhua Li
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuedong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shengnan Ren
- Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
4
|
Sebastião AI, Simões G, Oliveira F, Mateus D, Falcão A, Carrascal MA, Gomes C, Neves B, Cruz MT. Dendritic cells in triple-negative breast cancer: From pathophysiology to therapeutic applications. Cancer Treat Rev 2025; 133:102884. [PMID: 39837068 DOI: 10.1016/j.ctrv.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025]
Abstract
Breast cancer is the second most commonly diagnosed cancer in women and the fifth leading cause of cancer-related deaths worldwide. It is a highly heterogeneous disease, consisting of multiple subtypes that vary significantly in clinical characteristics and survival outcomes. Triple-negative breast cancer (TNBC) is a particularly aggressive and challenging subtype of breast cancer. Several immunotherapeutic approaches have been tested in patients with TNBC to improve disease outcomes, including the administration of dendritic cell (DC)-based vaccines. DCs are a heterogeneous cell population that play a crucial role in bridging the innate and adaptive immune systems. Therefore, DCs have been increasingly used in cancer vaccines due to their ability to prime and boost antigen specific T-cell immune responses. This review aims to provide a comprehensive overview of TNBC, including potential targets and pharmacological strategies, as well as an overview of DCs and their relevance in TNBC. In addition, we review ongoing clinical trials and shed light on the evolving landscape of DC-based immunotherapy for TNBC.
Collapse
Affiliation(s)
- Ana Isabel Sebastião
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gonçalo Simões
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Filomena Oliveira
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Mateus
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; BioMark@UC/CEB-LABBELS, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
| | | | - Célia Gomes
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research - iCBR, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bruno Neves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
5
|
Graciotti M, Kandalaft LE. Vaccines for cancer prevention: exploring opportunities and navigating challenges. Nat Rev Drug Discov 2025; 24:134-150. [PMID: 39622986 DOI: 10.1038/s41573-024-01081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 02/06/2025]
Abstract
Improved understanding of cancer immunology has gradually brought increasing attention towards cancer-preventive vaccines as an important tool in the fight against cancer. The aim of this approach is to reduce cancer occurrence by inducing a specific immune response targeting tumours at an early stage before they can fully develop. The great advantage of preventive cancer vaccines lies in the potential to harness a less-compromised immune system in vaccine recipients before their immune responses become affected by the advanced status of the disease itself or by aggressive treatments such as chemotherapy. Successful implementation of immunoprevention against oncogenic viruses such as hepatitis B and papillomavirus has led to a dramatic decrease in virally induced cancers. Extending this approach to other cancers holds great promise but remains a major challenge. Here, we provide a comprehensive review of preclinical evidence supporting this approach, encouraging results from pioneering clinical studies as well as a discussion on the key aspects and open questions to address in order to design potent prophylactic cancer vaccines in the near future.
Collapse
Affiliation(s)
- Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Lana E Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
- Department of Oncology, University of Lausanne (UNIL), Lausanne, Switzerland.
- AGORA Cancer Research Center, Lausanne, Lausanne, Switzerland.
- Swiss Medical Network, Genolier Innovation Network, Genolier Clinic, Genolier, Switzerland.
| |
Collapse
|
6
|
Hu Y, Wang C, Liang H, Li J, Yang Q. The treatment landscape of triple-negative breast cancer. Med Oncol 2024; 41:236. [PMID: 39210220 DOI: 10.1007/s12032-024-02456-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Triple-negative breast cancer (TNBC) tumors are biologically aggressive breast cancer. On the molecular level, TNBC is a highly heterogeneous disease; more biotechnologies are gradually being used to advance the understanding of TNBC subtypes and help establish more targeted therapies. Multiple TNBC target-related agents are already approved by the Food and Drug Administration for clinical use, including PI3K/AKT/mTOR inhibitors, PRAP inhibitors, and antibody-drug conjugates. Some innovative approaches, like peptide strategies, also promise to treat TNBC. Currently, the interplay between TNBC tumors and their tumor microenvironment provides a promising prospect for improving the efficacy of immunotherapy. In this review, we summarize the prevalent TNBC subtype methodologies, discuss the evolving therapeutic strategies, and propose new therapeutic possibilities based on existing foundational theories, with the attempt to serve as a reference to further advance tailoring treatment of TNBC.
Collapse
Affiliation(s)
- Yi Hu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Chen Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Huishi Liang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Jie Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China.
| | - Qiong Yang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China.
| |
Collapse
|
7
|
He YR, Xia H, Yun P, Xu Y, Ma WMJ, Xiao ZX, Zha GF. α-Lactalbumin mRNA-LNP Evokes an Anti-Tumor Effect Combined with Surgery in Triple-Negative Breast Cancer. Pharmaceutics 2024; 16:940. [PMID: 39065637 PMCID: PMC11279974 DOI: 10.3390/pharmaceutics16070940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) has been considered a huge clinical unmet need due to its aggressive progression and highly frequent metastasis. mRNA therapeutics supply a potential and versatile immunotherapy of oncology treatment. Here, we developed α-lactalbumin mRNA-lipid nanoparticles (α-LNP) as a potential therapeutical strategy for TNBC. The α-LNP induced the specific IgG antibodies and activated IFN γ-secreting-T cells in vivo. Additionally, the safety of α-LNP also had been demonstrated in vivo. When vaccinated prior to tumor implantation, α-LNP showed a preventive effect against 4T1 tumor growth and extended the survival of the tumor model by activating the memory immune responses. Furthermore, α-LNP administration in combination with surgical removal of neoplasm effectively inhibited the progression and metastasis in the TNBC model. Taken together, our results indicate that the α-LNP vaccine is a promising novel treatment for both therapeutics and prophylactics in TNBC.
Collapse
Affiliation(s)
- Yun-Ru He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Heng Xia
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Peng Yun
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China;
| | - Yuandong Xu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Winson M. J. Ma
- Shenzhen Institute of Advanced Technology, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China;
| | - Ze-Xiu Xiao
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen 518107, China
| | - Gao-Feng Zha
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| |
Collapse
|
8
|
Houlahan KE, Khan A, Greenwald NF, Vivas CS, West RB, Angelo M, Curtis C. Germline-mediated immunoediting sculpts breast cancer subtypes and metastatic proclivity. Science 2024; 384:eadh8697. [PMID: 38815010 DOI: 10.1126/science.adh8697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/05/2024] [Indexed: 06/01/2024]
Abstract
Tumors with the same diagnosis can have different molecular profiles and response to treatment. It remains unclear when and why these differences arise. Somatic genomic aberrations occur within the context of a highly variable germline genome. Interrogating 5870 breast cancer lesions, we demonstrated that germline-derived epitopes in recurrently amplified genes influence somatic evolution by mediating immunoediting. Individuals with a high germline-epitope burden in human epidermal growth factor receptor 2 (HER2/ERBB2) are less likely to develop HER2-positive breast cancer compared with other subtypes. The same holds true for recurrent amplicons defining three aggressive estrogen receptor (ER)-positive subgroups. Tumors that overcome such immune-mediated negative selection are more aggressive and demonstrate an "immune cold" phenotype. These data show that the germline genome plays a role in dictating somatic evolution.
Collapse
Affiliation(s)
- Kathleen E Houlahan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Noah F Greenwald
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Christina Curtis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
9
|
Gautam N, Ramamoorthi G, Champion N, Han HS, Czerniecki BJ. Reviewing the significance of dendritic cell vaccines in interrupting breast cancer development. Mol Aspects Med 2024; 95:101239. [PMID: 38150884 DOI: 10.1016/j.mam.2023.101239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
Breast cancer is a heterogeneous disease and is the most prevalent cancer in women. According to the U.S breast cancer statistics, about 1 in every 8 women develop an invasive form of breast cancer during their lifetime. Immunotherapy has been a significant advancement in the treatment of cancer with multiple studies reporting favourable patient outcomes by modulating the immune response to cancer cells. Here, we review the significance of dendritic cell vaccines in treating breast cancer patients. We discuss the involvement of dendritic cells and oncodrivers in breast tumorigenesis, highlighting the rationale for targeting oncodrivers and neoantigens using dendritic cell vaccine therapy. We review different dendritic cell subsets and maturation states previously used to develop vaccines and suggest the use of DC vaccines for breast cancer prevention. Further, we highlight that the intratumoral delivery of type 1 dendritic cell vaccines in breast cancer patients activates tumor antigen-specific CD4+ T helper cell type 1 (Th1) cells, promoting an anti-tumorigenic immune response while concurrently blocking pro-tumorigenic responses. In summary, this review provides an overview of the current state of dendritic cell vaccines in breast cancer highlighting the challenges and considerations necessary for an efficient dendritic cell vaccine design in interrupting breast cancer development.
Collapse
Affiliation(s)
- Namrata Gautam
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Ganesan Ramamoorthi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Nicholas Champion
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Hyo S Han
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Brian J Czerniecki
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
10
|
Liu X, Cheng Y, Mu Y, Zhang Z, Tian D, Liu Y, Hu X, Wen T. Diverse drug delivery systems for the enhancement of cancer immunotherapy: an overview. Front Immunol 2024; 15:1328145. [PMID: 38298192 PMCID: PMC10828056 DOI: 10.3389/fimmu.2024.1328145] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Despite the clear benefits demonstrated by immunotherapy, there is still an inevitable off-target effect resulting in serious adverse immune reactions. In recent years, the research and development of Drug Delivery System (DDS) has received increased prominence. In decades of development, DDS has demonstrated the ability to deliver drugs in a precisely targeted manner to mitigate side effects and has the advantages of flexible control of drug release, improved pharmacokinetics, and drug distribution. Therefore, we consider that combining cancer immunotherapy with DDS can enhance the anti-tumor ability. In this paper, we provide an overview of the latest drug delivery strategies in cancer immunotherapy and briefly introduce the characteristics of DDS based on nano-carriers (liposomes, polymer nano-micelles, mesoporous silica, extracellular vesicles, etc.) and coupling technology (ADCs, PDCs and targeted protein degradation). Our aim is to show readers a variety of drug delivery platforms under different immune mechanisms, and analyze their advantages and limitations, to provide more superior and accurate targeting strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yao Mu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | | | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Zaluzec EK, Sempere LF. Systemic and Local Strategies for Primary Prevention of Breast Cancer. Cancers (Basel) 2024; 16:248. [PMID: 38254741 PMCID: PMC10814018 DOI: 10.3390/cancers16020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
One in eight women will develop breast cancer in the US. For women with moderate (15-20%) to average (12.5%) risk of breast cancer, there are few options available for risk reduction. For high-risk (>20%) women, such as BRCA mutation carriers, primary prevention strategies are limited to evidence-based surgical removal of breasts and/or ovaries and anti-estrogen treatment. Despite their effectiveness in risk reduction, not many high-risk individuals opt for surgical or hormonal interventions due to severe side effects and potentially life-changing outcomes as key deterrents. Thus, better communication about the benefits of existing strategies and the development of new strategies with minimal side effects are needed to offer women adequate risk-reducing interventions. We extensively review and discuss innovative investigational strategies for primary prevention. Most of these investigational strategies are at the pre-clinical stage, but some are already being evaluated in clinical trials and others are expected to lead to first-in-human clinical trials within 5 years. Likely, these strategies would be initially tested in high-risk individuals but may be applicable to lower-risk women, if shown to decrease risk at a similar rate to existing strategies, but with minimal side effects.
Collapse
Affiliation(s)
- Erin K. Zaluzec
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pharmacology & Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Lorenzo F. Sempere
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
12
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
13
|
Liu Y, Hu Y, Xue J, Li J, Yi J, Bu J, Zhang Z, Qiu P, Gu X. Advances in immunotherapy for triple-negative breast cancer. Mol Cancer 2023; 22:145. [PMID: 37660039 PMCID: PMC10474743 DOI: 10.1186/s12943-023-01850-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Immunotherapy has recently emerged as a treatment strategy which stimulates the human immune system to kill tumor cells. Tumor immunotherapy is based on immune editing, which enhances the antigenicity of tumor cells and increases the tumoricidal effect of immune cells. It also suppresses immunosuppressive molecules, activates or restores immune system function, enhances anti-tumor immune responses, and inhibits the growth f tumor cell. This offers the possibility of reducing mortality in triple-negative breast cancer (TNBC). MAIN BODY Immunotherapy approaches for TNBC have been diversified in recent years, with breakthroughs in the treatment of this entity. Research on immune checkpoint inhibitors (ICIs) has made it possible to identify different molecular subtypes and formulate individualized immunotherapy schedules. This review highlights the unique tumor microenvironment of TNBC and integrates and analyzes the advances in ICI therapy. It also discusses strategies for the combination of ICIs with chemotherapy, radiation therapy, targeted therapy, and emerging treatment methods such as nanotechnology, ribonucleic acid vaccines, and gene therapy. Currently, numerous ongoing or completed clinical trials are exploring the utilization of immunotherapy in conjunction with existing treatment modalities for TNBC. The objective of these investigations is to assess the effectiveness of various combined immunotherapy approaches and determine the most effective treatment regimens for patients with TNBC. CONCLUSION This review provides insights into the approaches used to overcome drug resistance in immunotherapy, and explores the directions of immunotherapy development in the treatment of TNBC.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yueting Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jingying Li
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
14
|
Ghanta KP, Bandyopadhyay S. Counteraction Effects of Ammonium-Based Ionic Liquids on Urea-Induced Denaturation of α-Lactalbumin: A Comprehensive Molecular Simulation Study. J Phys Chem B 2023; 127:7251-7265. [PMID: 37574910 DOI: 10.1021/acs.jpcb.3c03223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Ionic liquids (ILs) are known to stabilize protein conformations in aqueous medium. Importantly, ILs can also act as refolding additives in urea-driven denaturation of proteins. However, despite the importance of the problem, detailed microscopic understanding of the counteraction effects of ILs on urea-induced protein denaturation remains elusive. In this work, atomistic molecular dynamics (MD) simulations of the protein α-lactalbumin have been carried out in pure aqueous medium, in 8 M binary urea-water solution and in ternary urea-IL-water solutions containing ammonium-based ethyl ammonium acetate (EAA) as the IL at different concentrations (1-4 M). Attempts have been made to quantify detailed molecular-level understanding of the origin behind the counteraction effects of the IL on urea-induced partial unfolding of the protein. The calculations revealed significant conformational changes of the protein with multiple free energy minima due to its partial unfolding in binary urea-water solution. The counteraction effect of the IL was evident from the enhanced structural rigidity of the protein with propensity to transform into a single native free energy minimum state in ternary urea-IL-water solutions. Such an effect has been found to be associated with preferential direct binding of the IL components with the protein and simultaneous expulsion of urea from the interface, thereby providing additional stabilization of the protein in ternary solutions. Most importantly, modified rearrangement of the hydrogen bond network at the interface due to the formation of stronger protein-cation (PC) and protein-anion (PA) hydrogen bonds by breaking relatively weaker protein-urea (PU) and protein-water (PW) hydrogen bonds has been recognized as the microscopic origin behind the counteraction effects of EAA on urea-induced partial unfolding of the protein.
Collapse
Affiliation(s)
- Krishna Prasad Ghanta
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
15
|
Zhang M, Huang Y, Zou J, Yang Y, Yao Y, Cheng G, Yang Y. Advanced Oxidation Nanoprocessing Boosts Immunogenicity of Whole Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302250. [PMID: 37211712 PMCID: PMC10401122 DOI: 10.1002/advs.202302250] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/11/2023] [Indexed: 05/23/2023]
Abstract
Whole tumor cells expressing a wide array of tumor antigens are considered as a highly promising source of antigens for cancer vaccines. However, simultaneously preserving the antigen diversity, improving immunogenicity, and eliminating the potential tumorigenic risk of whole tumor cells are highly challenging. Inspired by the recent progress in sulfate radical-based environmental technology, herein, an advanced oxidation nanoprocessing (AONP) strategy is developed for boosting the immunogenicity of whole tumor cells. The AONP is based on the activation of peroxymonosulfate by ZIF-67 nanocatalysts to produce SO4 -∙ radicals continuously, leading to sustained oxidative damage to tumor cells and consequently extensive cell death. Importantly, AONP causes immunogenic apoptosis as evidenced by the release of a series of characteristic damage associated molecular patterns and at the same time maintains the integrity of cancer cells, which is critical to preserve the cellular components and thus maximize the diversity of antigens. Finally, the immunogenicity of AONP-treated whole tumor cells is evaluated in a prophylactic vaccination model, demonstrating significantly delayed tumor growth and increased survival rate of live tumor-cell-challenged mice. It is expected that the developed AONP strategy would pave the way to develop effective personalized whole tumor cell vaccines in future.
Collapse
Affiliation(s)
- Min Zhang
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghai200092P. R. China
| | - Yiming Huang
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghai200092P. R. China
| | - Jie Zou
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghai200092P. R. China
| | - Yang Yang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of EducationSchool of Chemistry and Chemical EngineeringShandong UniversityJinanShandong250100P. R. China
| | - Yue Yao
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghai200092P. R. China
| | - Guofeng Cheng
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghai200092P. R. China
| | - Yannan Yang
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of OptoelectronicsFudan UniversityShanghai200433P. R. China
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueensland4072Australia
| |
Collapse
|
16
|
Houlahan KE, Khan A, Greenwald NF, West RB, Angelo M, Curtis C. Germline-mediated immunoediting sculpts breast cancer subtypes and metastatic proclivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532870. [PMID: 36993286 PMCID: PMC10055121 DOI: 10.1101/2023.03.15.532870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cancer represents a broad spectrum of molecularly and morphologically diverse diseases. Individuals with the same clinical diagnosis can have tumors with drastically different molecular profiles and clinical response to treatment. It remains unclear when these differences arise during disease course and why some tumors are addicted to one oncogenic pathway over another. Somatic genomic aberrations occur within the context of an individual's germline genome, which can vary across millions of polymorphic sites. An open question is whether germline differences influence somatic tumor evolution. Interrogating 3,855 breast cancer lesions, spanning pre-invasive to metastatic disease, we demonstrate that germline variants in highly expressed and amplified genes influence somatic evolution by modulating immunoediting at early stages of tumor development. Specifically, we show that the burden of germline-derived epitopes in recurrently amplified genes selects against somatic gene amplification in breast cancer. For example, individuals with a high burden of germline-derived epitopes in ERBB2, encoding human epidermal growth factor receptor 2 (HER2), are significantly less likely to develop HER2-positive breast cancer compared to other subtypes. The same holds true for recurrent amplicons that define four subgroups of ER-positive breast cancers at high risk of distant relapse. High epitope burden in these recurrently amplified regions is associated with decreased likelihood of developing high risk ER-positive cancer. Tumors that overcome such immune-mediated negative selection are more aggressive and demonstrate an "immune cold" phenotype. These data show the germline genome plays a previously unappreciated role in dictating somatic evolution. Exploiting germline-mediated immunoediting may inform the development of biomarkers that refine risk stratification within breast cancer subtypes.
Collapse
Affiliation(s)
- Kathleen E. Houlahan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah F Greenwald
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert B. West
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Curtis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
17
|
Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, Qiao B, Wang C. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther 2023; 8:124. [PMID: 36922504 PMCID: PMC10017761 DOI: 10.1038/s41392-023-01382-y] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/31/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Exosome is a subgroup of extracellular vesicles, which has been serving as an efficient therapeutic tool for various diseases. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. After appropriate modification, engineered exosomes are able to deliver antitumor drugs to tumor sites efficiently and precisely with fewer treatment-related adverse effects. However, there still exist many challenges for the clinical translation of engineered exosomes. For instance, what sources and modification strategies could endow exosomes with the most efficient antitumor activity is still poorly understood. Additionally, how to choose appropriately engineered exosomes in different antitumor therapies is another unresolved problem. In this review, we summarized the characteristics of engineered exosomes, especially the spatial and temporal properties. Additionally, we concluded the recent advances in engineered exosomes in the cancer fields, including the sources, isolation technologies, modification strategies, and labeling and imaging methods of engineered exosomes. Furthermore, the applications of engineered exosomes in different antitumor therapies were summarized, such as photodynamic therapy, gene therapy, and immunotherapy. Consequently, the above provides the cancer researchers in this community with the latest ideas on engineered exosome modification and new direction of new drug development, which is prospective to accelerate the clinical translation of engineered exosomes for cancer-targeted therapy.
Collapse
Affiliation(s)
- Menghui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Bingbing Qiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
18
|
Husseini RA, Abe N, Hara T, Abe H, Kogure K. Use of Iontophoresis Technology for Transdermal Delivery of a Minimal mRNA Vaccine as a Potential Melanoma Therapeutic. Biol Pharm Bull 2023; 46:301-308. [PMID: 36724958 DOI: 10.1248/bpb.b22-00746] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
mRNA vaccines have attracted considerable attention as a result of the 2019 coronavirus pandemic; however, challenges remain regarding use of mRNA vaccines, including insufficient delivery owing to the high molecular weights and high negative charges associated with mRNA. These characteristics of mRNA vaccines impair intracellular uptake and subsequent protein translation. In the current study, we prepared a minimal mRNA vaccine encoding a tumor associated antigen human gp10025-33 peptide (KVPRNQDWL), as a potential treatment for melanoma. Minimal mRNA vaccines have recently shown promise at improving the translational process, and can be prepared via a simple production method. Moreover, we previously reported the successful use of iontophoresis (IP) technology in the delivery of hydrophilic macromolecules into skin layers, as well as intracellular delivery of small interfering RNA (siRNA). We hypothesized that combining IP technology with a newly synthesized minimal mRNA vaccine can improve both transdermal and intracellular delivery of mRNA. Following IP-induced delivery of a mRNA vaccine, an immune response is elicited resulting in activation of skin resident immune cells. As expected, combining both technologies led to potent stimulation of the immune system, which was observed via potent tumor inhibition in mice bearing melanoma. Additionally, there was an elevation in mRNA expression levels of various cytokines, mainly interferon (IFN)-γ, as well as infiltration of cytotoxic CD8+ T cells in the tumor tissue, which are responsible for tumor clearance. This is the first report demonstrating the application of IP for delivery of a minimal mRNA vaccine as a potential melanoma therapeutic.
Collapse
Affiliation(s)
- Rabab A Husseini
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University.,Department of Pharmaceutical Health Chemistry, Graduate School of Pharmaceutical Sciences, Tokushima University
| | - Naoko Abe
- Department of Chemistry, Graduate School of Science, Nagoya University
| | - Tomoaki Hara
- Department of Chemistry, Graduate School of Science, Nagoya University
| | - Hiroshi Abe
- Department of Chemistry, Graduate School of Science, Nagoya University
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University
| |
Collapse
|
19
|
Cancer Vaccines for Triple-Negative Breast Cancer: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11010146. [PMID: 36679991 PMCID: PMC9866612 DOI: 10.3390/vaccines11010146] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the subtype of breast cancer with the poorest outcomes, and is associated with a high risk of relapse and metastasis. The treatment choices for this malignancy have been confined to conventional chemotherapeutic agents, due to a lack of expression of the canonical molecular targets. Immunotherapy has been recently changing the treatment paradigm for many types of tumors, and the approach of evoking active immune responses in the milieu of breast tumors through cancer vaccines has been introduced as one of the most novel immunotherapeutic approaches. Accordingly, a number of vaccines for the treatment or prevention of recurrence have been developed and are currently being studied in TNBC patients, while none have yet received any approvals. To elucidate the efficacy and safety of these vaccines, we performed a systematic review of the available literature on the topic. After searching the PubMed, Scopus, Web of Science, Embase, Cochrane CENTRAL, and Google Scholar databases, a total of 5701 results were obtained, from which 42 clinical studies were eventually included based on the predefined criteria. The overall quality of the included studies was acceptable. However, due to a lack of reporting outcomes of survival or progression in some studies (which were presented as conference abstracts) as well as the heterogeneity of the reported outcomes and study designs, we were not able to carry out a meta-analysis. A total of 32 different vaccines have so far been evaluated in TNBC patients, with the majority belonging to the peptide-based vaccine type. The other vaccines were in the cell or nucleic acid (RNA/DNA)-based categories. Most vaccines proved to be safe with low-grade, local adverse events and could efficiently evoke cellular immune responses; however, most trials were not able to demonstrate significant improvements in clinical indices of efficacy. This is in part due to the limited number of randomized studies, as well as the limited TNBC population of each trial. However, due to the encouraging results of the currently published trials, we anticipate that this strategy could show its potential through larger, phase III randomized studies in the near future.
Collapse
|
20
|
Chen G, Xiong W, Gu Z, Gao Y, Hou J, Long L, Wang H, Asrorov AM, Muhitdinov B, Xu Q, Huang Y. Mannosylated engineered trichosanthin-legumain protein vaccine hydrogel for breast cancer immunotherapy. Int J Biol Macromol 2022; 223:1485-1494. [PMID: 36395942 DOI: 10.1016/j.ijbiomac.2022.11.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/27/2022] [Accepted: 11/06/2022] [Indexed: 11/16/2022]
Abstract
The development of cancer vaccines based on tumor-associated antigens is hurdled by lack of an efficient adjuvant and insufficient efficacy. To improve the efficacy of vaccines, a genetically-engineered method was employed in this work to achieve the codelivery of antigen and adjuvant to enhance immune responses. Trichosanthin is a plant-derived protein that possesses cancer immune stimulation function. A genetically engineered protein vaccine composed of trichosanthin (adjuvant) and legumain domain (a peptidic antigen) was constructed, which was further chemically modified with mannose for targeting dendritic cells (DCs). The method is facile and ready for scaling up for massive production. Such a "two-in-one" vaccine is advantageous for codelivery for augmenting the immune responses. The vaccine inhibited the tumors by triggering a robust cytotoxic T lymphocyte response in the orthotopic-breast-tumor mice. Furthermore, the vaccine was loaded into the temperature-sensitive hydrogel based on Pluronic F127 for implanting use in the post-surgical site. The sustained-released vaccine from the hydrogel inhibited not only the tumor recurrence but also the lung metastases of breast cancer. These findings demonstrated that it was a safe and effective vaccination for breast cancer immunotherapy in a prophylactical and therapeutical manner for remodeling the tumor immune microenvironment and arresting tumor growth.
Collapse
Affiliation(s)
- Guihua Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510450, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, CAS, Zhongshan 528437, China
| | - Wei Xiong
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510450, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, CAS, Zhongshan 528437, China
| | - Zeyun Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Yanrong Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Jiazhen Hou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, 10, Nanjing 210023, China
| | - Li Long
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510450, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, CAS, Zhongshan 528437, China
| | - Huiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Bahtiyor Muhitdinov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510450, China.
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, CAS, Zhongshan 528437, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, 10, Nanjing 210023, China.
| |
Collapse
|
21
|
Zhang X, Cui H, Zhang W, Li Z, Gao J. Engineered tumor cell-derived vaccines against cancer: The art of combating poison with poison. Bioact Mater 2022; 22:491-517. [PMID: 36330160 PMCID: PMC9619151 DOI: 10.1016/j.bioactmat.2022.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccination is a promising approach for tumor immunotherapy because it presents high specificity and few side effects. However, tumor vaccines that contain only a single tumor antigen can allow immune system evasion by tumor variants. Tumor antigens are complex and heterogeneous, and identifying a single antigen that is uniformly expressed by tumor cells is challenging. Whole tumor cells can produce comprehensive antigens that trigger extensive tumor-specific immune responses. Therefore, tumor cells are an ideal source of antigens for tumor vaccines. A better understanding of tumor cell-derived vaccines and their characteristics, along with the development of new technologies for antigen delivery, can help improve vaccine design. In this review, we summarize the recent advances in tumor cell-derived vaccines in cancer immunotherapy and highlight the different types of engineered approaches, mechanisms, administration methods, and future perspectives. We discuss tumor cell-derived vaccines, including whole tumor cell components, extracellular vesicles, and cell membrane-encapsulated nanoparticles. Tumor cell-derived vaccines contain multiple tumor antigens and can induce extensive and potent tumor immune responses. However, they should be engineered to overcome limitations such as insufficient immunogenicity and weak targeting. The genetic and chemical engineering of tumor cell-derived vaccines can greatly enhance their targeting, intelligence, and functionality, thereby realizing stronger tumor immunotherapy effects. Further advances in materials science, biomedicine, and oncology can facilitate the clinical translation of tumor cell-derived vaccines.
Collapse
Affiliation(s)
- Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Hengqing Cui
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Corresponding author. Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Corresponding author. Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200444, China.
| |
Collapse
|
22
|
Huang L, Rong Y, Tang X, Yi K, Qi P, Hou J, Liu W, He Y, Gao X, Yuan C, Wang F. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Mol Cancer 2022; 21:45. [PMID: 35148751 PMCID: PMC8831689 DOI: 10.1186/s12943-022-01515-x] [Citation(s) in RCA: 221] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022] Open
Abstract
Background Dendritic cells (DCs) are central for the initiation and regulation of innate and adaptive immunity in the tumor microenvironment. As such, many kinds of DC-targeted vaccines have been developed to improve cancer immunotherapy in numerous clinical trials. Targeted delivery of antigens and adjuvants to DCs in vivo represents an important approach for the development of DC vaccines. However, nonspecific activation of systemic DCs and the preparation of optimal immunodominant tumor antigens still represent major challenges. Methods We loaded the immunogenic cell death (ICD) inducers human neutrophil elastase (ELANE) and Hiltonol (TLR3 agonist) into α-lactalbumin (α-LA)-engineered breast cancer-derived exosomes to form an in situ DC vaccine (HELA-Exos). HELA-Exos were identified by transmission electron microscopy, nanoscale flow cytometry, and Western blot analysis. The targeting, killing, and immune activation effects of HELA-Exos were evaluated in vitro. The tumor suppressor and immune-activating effects of HELA-Exos were explored in immunocompetent mice and patient-derived organoids. Results HELA-Exos possessed a profound ability to specifically induce ICD in breast cancer cells. Adequate exposure to tumor antigens and Hiltonol following HELA-Exo-induced ICD of cancer cells activated type one conventional DCs (cDC1s) in situ and cross-primed tumor-reactive CD8+ T cell responses, leading to potent tumor inhibition in a poorly immunogenic triple negative breast cancer (TNBC) mouse xenograft model and patient-derived tumor organoids. Conclusions HELA-Exos exhibit potent antitumor activity in both a mouse model and human breast cancer organoids by promoting the activation of cDC1s in situ and thus improving the subsequent tumor-reactive CD8+ T cell responses. The strategy proposed here is promising for generating an in situ DC-primed vaccine and can be extended to various types of cancers. Graphic Abstract Scheme 1. Schematic illustration of HELA-Exos as an in situ DC-primed vaccine for breast cancer. (A) Allogenic breast cancer-derived exosomes isolated from MDA-MB-231 cells were genetically engineered to overexpress α-LA and simultaneously loaded with the ICD inducers ELANE and Hiltonol (TLR3 agonist) to generate HELA-Exos. (B) Mechanism by which HELA-Exos activate DCs in situ in a mouse xenograft model ofTNBC. HELA-Exos specifically homed to the TME and induced ICD in cancer cells, which resulted in the increased release of tumor antigens, Hiltonol, and DAMPs, as well as the uptake of dying tumor cells by cDC1s. The activated cDC1s then cross-primed tumor-reactive CD8+ T cell responses. (C) HELA-Exos activated DCs in situ in the breast cancer patient PBMC-autologous tumor organoid coculture system. Abbreviations: DCs: dendritic cells; α-LA: α-lactalbumin; HELA-Exos: Hiltonol-ELANE-α-LA-engineered exosomes; ICD: immunogenic cell death; ELANE: human neutrophil elastase; TLR3: Toll-like receptor 3; TNBC: triple-negative breast cancer; TME: tumor microenvironment; DAMPs: damage-associated molecular patterns; cDC1s: type 1 conventional dendritic cells; PBMCs: peripheral blood mononuclear cells ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01515-x.
Collapse
Affiliation(s)
- Lanxiang Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, Wuchang District, China.,Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Rong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, Wuchang District, China.,Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Tang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, Wuchang District, China.,Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kezhen Yi
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, Wuchang District, China.,Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Qi
- Department of Thyroid and Breast Surgery, Hubei No. 3 People's Hospital of Jianghan University, Wuhan, China
| | - Jinxuan Hou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weihuang Liu
- Medical Research Center for Structural Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yuan He
- Medical Research Center for Structural Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xing Gao
- Animal Experiment Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunhui Yuan
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, Wuchang District, China. .,Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China. .,Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
23
|
Hu Y, Burkard ME. Classes of therapeutics to amplify the immune response. Breast Cancer Res Treat 2022; 191:277-289. [PMID: 34787761 PMCID: PMC11646685 DOI: 10.1007/s10549-021-06369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/18/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Conventional chemotherapies are a mainstay for metastatic breast cancers, though durable response is rare. Immunotherapies promise long-term responses thorough immune activation but have been underwhelming in breast cancer relative to other cancer types. Here, we review the mechanisms of existing strategies including chemotherapies and how they may cause breast cancers to become immunogenic to identify potential biomarkers for combinations of conventional and immunotherapies. CONCLUSION Mechanistic considerations should inform biomarker development and patient selection for therapeutic combinations of drugs to combine with immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Yang Hu
- Department of Medicine, Hematology/Oncology and Palliative Care, University of Wisconsin-Madison, Madison, WI, 53705, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mark E Burkard
- Department of Medicine, Hematology/Oncology and Palliative Care, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Wisconsin Institutes for Medical Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
24
|
Zachariah NN, Basu A, Gautam N, Ramamoorthi G, Kodumudi KN, Kumar NB, Loftus L, Czerniecki BJ. Intercepting Premalignant, Preinvasive Breast Lesions Through Vaccination. Front Immunol 2021; 12:786286. [PMID: 34899753 PMCID: PMC8652247 DOI: 10.3389/fimmu.2021.786286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) prevention remains the ultimate cost-effective method to reduce the global burden of invasive breast cancer (IBC). To date, surgery and chemoprevention remain the main risk-reducing modalities for those with hereditary cancer syndromes, as well as high-risk non-hereditary breast lesions such as ADH, ALH, or LCIS. Ductal carcinoma in situ (DCIS) is a preinvasive malignant lesion of the breast that closely mirrors IBC and, if left untreated, develops into IBC in up to 50% of lesions. Certain high-risk patients with DCIS may have a 25% risk of developing recurrent DCIS or IBC, even after surgical resection. The development of breast cancer elicits a strong immune response, which brings to prominence the numerous advantages associated with immune-based cancer prevention over drug-based chemoprevention, supported by the success of dendritic cell vaccines targeting HER2-expressing BC. Vaccination against BC to prevent or interrupt the process of BC development remains elusive but is a viable option. Vaccination to intercept preinvasive or premalignant breast conditions may be possible by interrupting the expression pattern of various oncodrivers. Growth factors may also function as potential immune targets to prevent breast cancer progression. Furthermore, neoantigens also serve as effective targets for interception by virtue of strong immunogenicity. It is noteworthy that the immune response also needs to be strong enough to result in target lesion elimination to avoid immunoediting as it may occur in IBC arising from DCIS. Overall, if the issue of vaccine targets can be solved by interrupting premalignant lesions, there is a potential to prevent the development of IBC.
Collapse
Affiliation(s)
| | - Amrita Basu
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Namrata Gautam
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Krithika N Kodumudi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Nagi B Kumar
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Loretta Loftus
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J Czerniecki
- Department of Breast Surgery, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
25
|
Cancer Vaccines: Promising Therapeutics or an Unattainable Dream. Vaccines (Basel) 2021; 9:vaccines9060668. [PMID: 34207062 PMCID: PMC8233841 DOI: 10.3390/vaccines9060668] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 02/08/2023] Open
Abstract
The advent of cancer immunotherapy has revolutionized the field of cancer treatment and offers cancer patients new hope. Although this therapy has proved highly successful for some patients, its efficacy is not all encompassing and several cancer types do not respond. Cancer vaccines offer an alternate approach to promote anti-tumor immunity that differ in their mode of action from antibody-based therapies. Cancer vaccines serve to balance the equilibrium of the crosstalk between the tumor cells and the host immune system. Recent advances in understanding the nature of tumor-mediated tolerogenicity and antigen presentation has aided in the identification of tumor antigens that have the potential to enhance anti-tumor immunity. Cancer vaccines can either be prophylactic (preventative) or therapeutic (curative). An exciting option for therapeutic vaccines is the emergence of personalized vaccines, which are tailor-made and specific for tumor type and individual patient. This review summarizes the current standing of the most promising vaccine strategies with respect to their development and clinical efficacy. We also discuss prospects for future development of stem cell-based prophylactic vaccines.
Collapse
|
26
|
Keshavarz-Fathi M, Rezaei N. Cancer Immunoprevention: Current Status and Future Directions. Arch Immunol Ther Exp (Warsz) 2021; 69:3. [PMID: 33638703 DOI: 10.1007/s00005-021-00604-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/06/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most serious diseases affecting health and the second leading cause of death worldwide. Despite the development of various therapeutic modalities to deal with cancer, limited improvement in overall survival of patients has been yielded. Since there is no certain cure for cancer, detection of premalignant lesions, and prevention of their progression are vital to the decline of high morbidity and mortality of cancer. Among approaches to cancer prevention, immunoprevention has gained further attention in recent years. Deep understanding of the tumor/immune system interplay and successful prevention of virally-induced malignancies by vaccines have paved the way toward broadening cancer immunoprevention application. The identification of tumor antigens in premalignant lesions was the turning point in cancer immunoprevention that led to designing preventive vaccines for various malignancies including multiple myeloma, colorectal, and breast cancer. In addition to vaccines, immune checkpoint inhibitors are also being tested for the prevention of oral squamous cell carcinoma (SCC), and imiquimod which is an established drug for the prevention of skin SCC, is a non-specific immunomodulator. Herein, to provide a bench-to-bedside understanding of cancer immunoprevention, we will review the role of the immune system in suppression and promotion of tumors, immunoprevention of virally-induced cancers, identification of tumor antigens in premalignant lesions, and clinical advances of cancer immunoprevention.
Collapse
Affiliation(s)
- Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
27
|
Yang J, Zhao C, Lim J, Zhao L, Tourneau RL, Zhang Q, Dobson D, Joshi S, Pang J, Zhang X, Pal S, Andreou C, Zhang H, Kircher MF, Schmitthenner H. Structurally symmetric near-infrared fluorophore IRDye78-protein complex enables multimodal cancer imaging. Theranostics 2021; 11:2534-2549. [PMID: 33456558 PMCID: PMC7806473 DOI: 10.7150/thno.54928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: Most contemporary cancer therapeutic paradigms involve initial imaging as a treatment roadmap, followed by the active engagement of surgical operations. Current approved intraoperative contrast agents exemplified by indocyanine green (ICG) have a few drawbacks including the inability of pre-surgical localization. Alternative near-infrared (NIR) dyes including IRDye800cw are being explored in advanced clinical trials but often encounter low chemical yields and complex purifications owing to the asymmetric synthesis. A single contrast agent with ease of synthesis that works in multiple cancer types and simultaneously allows presurgical imaging, intraoperative deep-tissue three-dimensional visualization, and high-speed microscopic visualization of tumor margins via spatiotemporally complementary modalities would be beneficial. Methods: Due to the lack of commercial availability and the absence of detailed synthesis and characterization, we proposed a facile and scalable synthesis pathway for the symmetric NIR water-soluble heptamethine sulfoindocyanine IRDye78. The synthesis can be accomplished in four steps from commercially-available building blocks. Its symmetric resonant structure avoided asymmetric synthesis problems while still preserving the benefits of analogous IRDye800cw with commensurable optical properties. Next, we introduced a low-molecular-weight protein alpha-lactalbumin (α-LA) as the carrier that effectively modulates the hepatic clearance of IRDye78 into the preferred renal excretion pathway. We further implemented 89Zr radiolabeling onto the protein scaffold for positron emission tomography (PET). The multimodal imaging capability of the fluorophore-protein complex was validated in breast cancer and glioblastoma. Results: The scalable synthesis resulted in high chemical yields, typically 95% yield in the final step of the chloro dye. Chemical structures of intermediates and the final fluorophore were confirmed. Asymmetric IRDye78 exhibited comparable optical features as symmetric IRDye800cw. Its well-balanced quantum yield affords concurrent dual fluorescence and optoacoustic contrast without self-quenching nor concentration-dependent absorption. The NHS ester functionality modulates efficient covalent coupling to reactive side-chain amines to the protein carrier, along with desferrioxamine (DFO) for stable radiolabeling of 89Zr. The fluorophore-protein complex advantageously shifted the biodistribution and can be effectively cleared through the urinary pathway. The agent accumulates in tumors and enables triple-modal visualization in mouse xenograft models of both breast and brain cancers. Conclusion: This study described in detail a generalized strategic modulation of clearance routes towards the favorable renal clearance, via the introduction of α-LA. IRDye78 as a feasible alternative of IRDye800cw currently in clinical phases was proposed with a facile synthesis and fully characterized for the first time. This fluorophore-protein complex with stable radiolabeling should have great potential for clinical translation where it could enable an elegant workflow from preoperative planning to intraoperative deep tissue and high-resolution image-guided resection.
Collapse
|
28
|
Xin J, Zeng D, Wang H, Sun N, Khalique A, Zhao Y, Wu L, Pan K, Jing B, Ni X. Lactobacillus johnsonii BS15 improves intestinal environment against fluoride-induced memory impairment in mice-a study based on the gut-brain axis hypothesis. PeerJ 2020; 8:e10125. [PMID: 33083147 PMCID: PMC7547597 DOI: 10.7717/peerj.10125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/17/2020] [Indexed: 01/09/2023] Open
Abstract
Background Excessive fluoride can lead to chronic neurodegeneration characterized by neuron and myelin loss and memory dysfunction. The gut–brain axis hypothesis suggests that gut microbiota plays a crucial role in regulating brain function. Thus, using probiotics to adjust the gut microenvironment may be a potential therapy for mental diseases. Methods Mice in the prob group were administrated with Lactobacillus johnsonii BS15 for 28 days prior to and throughout a 70-day exposure to sodium fluoride. The drinking water of all groups (F and prob groups) except the control group were replaced by high-fluoride water (100 mg NaF/L) on day 28. Animals in each group were divided into two subsets: one underwent behavioral test, and the other was sacrificed for sampling. The mRNA expression level and protein content related to inflammatory reaction in the ileum and hippocampus were respectively detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). The mRNA expression levels of proteins related to myelin structure, apoptosis, and memory in the hippocampus and tight junction proteins in the ileum were determined by RT-qPCR and/or immunohistochemistry. Gut permeability markers (D-lactate and diamine oxidase (DAO)) in the serum were also examined by ELISA. Results The results showed that fluoride exposure induced a lower spontaneous exploration (P < 0.05) in T-maze test, which indicated an impairment of memory. Spontaneous exploration of BS15-treated mice was significantly higher (P < 0.05) than that in F group. Fluoride reduced (P < 0.05) levels of myelin structural protein (proteolipid protein) and neurogenesis-associated proteins (brain-derived neurotrophic factor and cAMP/Ca2+ responsive element-binding protein), induced disordered inflammatory cytokines (TNF-α, IFN-γ, and IL-6; P < 0.05), increased pro-apoptotic genes (caspase-3; P < 0.05), and decreased anti-apoptotic genes (Bcl-2; P < 0.05) in the hippocampus, of which the influences were reversed by BS15. BS15 treatment exerted significant preventive effects on reversing the gut inflammation induced by excessive fluoride intake by reducing (P < 0.05) the levels of pro-inflammatory cytokines (tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ)) and remarkably increasing (P < 0.05) the level of anti-inflammatory cytokines (IL-10). Moreover, the serum DAO activity and D-lactate concentration significantly increased by fluoride were also reduced (P < 0.05) by BS15. This result indicated the profitable effect of BS15 on gut permeability. Conclusion L. johnsonii BS15 intake could benefit the neuroinflammation and demyelination in the hippocampus by improving the gut environment and ameliorating fluorine-induced memory dysfunction.
Collapse
Affiliation(s)
- Jinge Xin
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hesong Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ying Zhao
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liqian Wu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Yang J, Wang T, Zhao L, Rajasekhar VK, Joshi S, Andreou C, Pal S, Hsu HT, Zhang H, Cohen IJ, Huang R, Hendrickson RC, Miele MM, Pei W, Brendel MB, Healey JH, Chiosis G, Kircher MF. Gold/alpha-lactalbumin nanoprobes for the imaging and treatment of breast cancer. Nat Biomed Eng 2020; 4:686-703. [PMID: 32661307 PMCID: PMC8255032 DOI: 10.1038/s41551-020-0584-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/11/2020] [Indexed: 02/03/2023]
Abstract
Theranostic agents should ideally be renally cleared and biodegradable. Here, we report the synthesis, characterization and theranostic applications of fluorescent ultrasmall gold quantum clusters that are stabilized by the milk metalloprotein alpha-lactalbumin. We synthesized three types of these nanoprobes that together display fluorescence across the visible and near-infrared spectra when excited at a single wavelength through optical colour coding. In live tumour-bearing mice, the near-infrared nanoprobe generates contrast for fluorescence, X-ray computed tomography and magnetic resonance imaging, and exhibits long circulation times, low accumulation in the reticuloendothelial system, sustained tumour retention, insignificant toxicity and renal clearance. An intravenously administrated near-infrared nanoprobe with a large Stokes shift facilitated the detection and image-guided resection of breast tumours in vivo using a smartphone with modified optics. Moreover, the partially unfolded structure of alpha-lactalbumin in the nanoprobe helps with the formation of an anti-cancer lipoprotein complex with oleic acid that triggers the inhibition of the MAPK and PI3K-AKT pathways, immunogenic cell death and the recruitment of infiltrating macrophages. The biodegradability and safety profile of the nanoprobes make them suitable for the systemic detection and localized treatment of cancer.
Collapse
Affiliation(s)
- Jiang Yang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tai Wang
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Lina Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | | | - Suhasini Joshi
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Chrysafis Andreou
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suchetan Pal
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsiao-Ting Hsu
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanwen Zhang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan J Cohen
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruimin Huang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbo Pei
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Matthew B Brendel
- Molecular Cytology Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Moritz F Kircher
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Ghanta KP, Pal T, Mondal S, Bandyopadhyay S. Microscopic Understanding of the Effect of Ionic Liquid on Protein from Molecular Simulation Studies. J Phys Chem B 2020; 124:3909-3921. [PMID: 32302476 DOI: 10.1021/acs.jpcb.0c02001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We have performed molecular dynamics (MD) simulations of the protein α-lactalbumin in aqueous solution containing the ionic liquid (IL) 1-butyl-3-methyl imidazolium tetrafluoroborate ([BMIM][BF4]) as the cosolvent at different concentrations. Attempts have been made to obtain quantitative understanding of the effects of the IL on the conformational features of the protein as well as the distributions of the IL and water around it. The calculations revealed enhanced rigidity of the protein with reduced conformational fluctuations and increasingly correlated local motions in the presence of the IL. Nonuniform relative population of the BMIM+ and BF4- ions at the protein surface with respect to that in the bulk solution has been observed. It is demonstrated that exchange of water by the IL around the protein results in rearrangement of the hydrogen bond network at the interface with breaking of protein-water hydrogen bonds and formation of protein-IL hydrogen bonds. Importantly, it is found that the protein forms increased number of stronger salt bridges in the presence of IL. This shows that the formation of a greater number of such stronger salt bridges is the origin behind the enhanced rigidity of the protein in the presence of the IL.
Collapse
Affiliation(s)
- Krishna Prasad Ghanta
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, India
| | - Tamisra Pal
- Centre for Computational and Data Sciences, Indian Institute of Technology, Kharagpur 721302, India
| | - Sandip Mondal
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, India.,Centre for Computational and Data Sciences, Indian Institute of Technology, Kharagpur 721302, India
| |
Collapse
|
31
|
Tuohy VK, Johnson JM, Mazumder S. Primary immunoprevention of adult onset cancers by vaccinating against retired tissue-specific self-proteins. Semin Immunol 2020; 47:101392. [PMID: 31926646 DOI: 10.1016/j.smim.2020.101392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/01/2020] [Indexed: 11/25/2022]
Abstract
Despite the enormous success of childhood prophylactic vaccination against diseases caused by pathogens, there is currently no similar preventive vaccine program against diseases confronted with age like breast cancer and ovarian cancer. With the exception of the annual influenza vaccine, current recommendations for adult vaccination are for either primary vaccines not received during childhood or for booster vaccinations to maintain the immunity against pathogens already induced during childhood. Here we describe a strategy to provide prophylactic pre-emptive immunity against the development of adult onset cancers not associated with any definitive etiopathogenic agent. We propose that safe and effective pre-emptive immunity may be induced in cancer-free subjects by vaccination against immunodominant tissue-specific self-proteins that are 'retired' from expression in normal tissues as part of the normal aging process but are expressed in tumors that emerge with age. Primary immunoprevention of adult onset cancers like breast cancer and ovarian cancer represents a great challenge and an even greater unmet need for our current healthcare.
Collapse
Affiliation(s)
- Vincent K Tuohy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA; Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH, USA.
| | - Justin M Johnson
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH, USA
| | - Suparna Mazumder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
32
|
Burke EE, Kodumudi K, Ramamoorthi G, Czerniecki BJ. Vaccine Therapies for Breast Cancer. Surg Oncol Clin N Am 2019; 28:353-367. [DOI: 10.1016/j.soc.2019.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Shoemaker RH, Forsthuber TG. Targeting "Retired Antigens" for Cancer Immunoprevention. Cancer Prev Res (Phila) 2019; 10:607-608. [PMID: 29093010 DOI: 10.1158/1940-6207.capr-17-0188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 11/16/2022]
Abstract
Identification of immune targets for cancer immunoprevention, or immunotherapy, has historically focused on tumor-associated (self) antigens or neoantigens expressed on malignant cells. For self-antigens, overcoming tolerance can be a difficult challenge. Neoantigens do not suffer from this limitation, but the lack of recurrent mutations yielding common neoantigens that can be exploited in vaccines is a problem for many tumor types. Targeting "retired antigens," a specialized type of self-antigen, may have considerable advantages. Antigens no longer expressed in mature or aged individuals should pose reduced risk of autoimmune sequelae. Indeed, self-tolerance of these antigens may have naturally faded. Thus, when the retired antigens are highly expressed in cancer cells, it may be easier to overcome the remaining tolerance. Women who are BRCA1/2 carriers may be among the first to benefit as candidate retired antigens have been identified as highly expressed in ovarian and breast cancer cells. Although there is good preclinical data supporting this immune targeting concept, additional research is needed to understand the underlying immune phenomena and optimize the vaccine strategy. Cancer Prev Res; 10(11); 607-8. ©2017 AACRSee related article by Mazumder et al., p. 612.
Collapse
Affiliation(s)
- Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Bethesda, Maryland.
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
34
|
Yaddanapudi K, Meng S, Whitt AG, Al Rayyan N, Richie J, Tu A, Eaton JW, Li C. Exosomes from GM-CSF expressing embryonic stem cells are an effective prophylactic vaccine for cancer prevention. Oncoimmunology 2019; 8:1561119. [PMID: 30723593 DOI: 10.1080/2162402x.2018.1561119] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/26/2018] [Accepted: 12/10/2018] [Indexed: 01/21/2023] Open
Abstract
The antigenic similarity between embryos and tumors has raised the idea of using embryonic material as a preventative vaccine against neoplastic disease. Indeed, we have previously reported that a vaccine comprises allogeneic murine embryonic stem cells (ESCs) and murine fibroblasts expressing GM-CSF (to amplify immune responses) successfully blocks the outgrowth of an implantable cancer (Lewis lung carcinoma; LLC) and lung tumors generated in mice using a combination of a mutagen followed by chronic pulmonary inflammation. However, such a vaccine is obviously impractical for application to humans. The use of fibroblasts to generate GM-CSF is needlessly complicated, and intact whole ESCs carry the hazard of generating embryomas/teratomas. Here, we report the successful application of an alternative prophylactic vaccine comprises exosomes derived from murine ESCs engineered to produce GM-CSF. Vaccination of mice with these exosomes significantly slowed or blocked the outgrowth of implanted LLC while control exosomes lacking GM-CSF were ineffective. Examination of tumor-infiltrating immune cells from mice vaccinated with the GM-CSF-expressing exosomes showed robust tumor-reactive CD8+ T effector responses, Th1 cytokine responses, and higher CD8+ T effector/CD4+CD25+Foxp3+ T regulatory cell ratio in the tumors. We conclude that a similar vaccine derived from GM-CSF- expressing human ESCs can be employed as a preventative vaccine for humans with an increased risk of developing cancer.
Collapse
Affiliation(s)
- Kavitha Yaddanapudi
- Immuno-Oncology Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.,Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Shuhan Meng
- Experimental Therapeutics Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Aaron G Whitt
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Experimental Therapeutics Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Numan Al Rayyan
- Immuno-Oncology Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jamaal Richie
- Immuno-Oncology Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Allison Tu
- Experimental Therapeutics Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - John W Eaton
- Immuno-Oncology Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Chi Li
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Experimental Therapeutics Program, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
35
|
Yaddanapudi K, Li C, Eaton JW. Vaccination with induced pluripotent stem cells confers protection against cancer. Stem Cell Investig 2018; 5:23. [PMID: 30148156 DOI: 10.21037/sci.2018.07.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/11/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Kavitha Yaddanapudi
- Molecular Targets Program, JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.,Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA.,Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Chi Li
- Molecular Targets Program, JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.,Department of Medicine, University of Louisville, Louisville, KY 40202, USA.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - John W Eaton
- Molecular Targets Program, JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
36
|
Jaini R, Loya MG, Eng C. Immunotherapeutic target expression on breast tumors can be amplified by hormone receptor antagonism: a novel strategy for enhancing efficacy of targeted immunotherapy. Oncotarget 2018; 8:32536-32549. [PMID: 28430646 PMCID: PMC5464807 DOI: 10.18632/oncotarget.15812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/18/2017] [Indexed: 11/30/2022] Open
Abstract
Immunotherapy has historically been successful in highly antigenic tumors but has shown limited therapeutic efficacy in non-antigenic tumors such as breast cancers. Our previous studies in autoimmunity have demonstrated that increased antigen load within a tissue enhances immune reactivity against it. We therefore hypothesized that enhancing expression of target proteins on breast tumors can increase efficacy of targeted immunotherapy. We hypothesized that antagonism of the estrogen receptor (ER) can increase expression of targets that are hormonally regulated and facilitate enhanced tumor recognition by targeted immunotherapy. We used a lactation protein α-Lactalbumin, a known immunotherapeutic target on breast tumors, as our model target antigen. Enhancement of target protein expression in human and murine breast tumors was tested in vitro and in vivo by ER antagonism using clinically established ER modulators, Tamoxifen and Fulvestrant. We show that antagonism of the ER can induce a 2–3 fold increase in expression of target proteins on tumors leaving the normal breast tissue unaffected. Tumor progression studies in 4T1 tumor-bearing mice show that efficacy of adoptively transferred cell based targeted immunotherapy was enhanced by target antigen amplification resulting in significantly higher tumor inhibition. However, in spite of increased target expression, anti-tumor efficacy of direct immunization was not enhanced probably due to other limiting factors involved in the immune priming process. Our study provides a novel combinatorial clinical strategy for enhancing efficacy of immunotherapy not only on breast tumors but potentially also for other hormonally driven tumors such as those of the prostate, testis and ovary.
Collapse
Affiliation(s)
- Ritika Jaini
- Genomic Medicine Institute, Lerner Research Institute, Cleveland, OH 44195, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Matthew G Loya
- Genomic Medicine Institute, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland, OH 44195, USA.,Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
37
|
Thomas F, Kareva I, Raven N, Hamede R, Pujol P, Roche B, Ujvari B. Evolved Dependence in Response to Cancer. Trends Ecol Evol 2018; 33:269-276. [DOI: 10.1016/j.tree.2018.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
|
38
|
Targeting the pro-angiogenic forms of VEGF or inhibiting their expression as anti-cancer strategies. Oncotarget 2018; 8:9174-9188. [PMID: 27999187 PMCID: PMC5354723 DOI: 10.18632/oncotarget.13942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
Tumor growth relies on oxygen and blood supply depending on neo-vascularization. This process is mediated by the Vascular Endothelial Growth Factor (VEGF) in many tumors. This paradigm has led to the development of specific therapeutic approaches targeting VEGF or its receptors. Despite their promising effects, these strategies have not improved overall survival of patients suffering from different cancers compared to standard therapies. We hypothesized that the existence of anti-angiogenic forms of VEGF VEGFxxxb which are still present in many tumors limit the therapeutic effects of the anti-VEGF antibodies bevacizumab/Avastin (BVZ). To test this hypothesis, we generated renal cell carcinoma cells (RCC) expressing VEGF165b. The incidence of tumors xenografts generated in nude mice and their growth were inferior to those obtained with control cells. Whereas BVZ had no effect on control tumors, it slowed-down the growth of tumor generated with VEGF165b expressing cells. A prophylactic immunization against the domain discriminating VEGF from VEGFxxxb isoforms inhibited the growth of tumor generated with two different syngenic tumor cell lines (melanoma (B16 cells) and RCC (RENCA cells)). Purified immunoglobulins from immunized mice also slowed-down tumor growth of human RCC xenografts in nude mice, producing a potent effect compared to BVZ in this model. Furthermore, down-regulating the serine-arginine-rich splicing factor 1 (SRSF1) or masking SRSF1 binding sites by 2'O-Methyl RNA resulted in the increase of the VEGFxxxb/VEGF ratio. Therefore, a vaccine approach, specific antibodies against pro-angiogenic forms of VEGF, or increasing the VEGFxxxb/VEGF ratio may represent new prophylactic or pro-active anti-cancer strategies.
Collapse
|
39
|
Abstract
Autoimmunity has both beneficial and harmful aspects. Beneficial aspects include: (1) removal of released intracytoplasmic antigens (ags) (cells at the end of their life span or damaged by outside agents) by specific nonpathogenic IgM autoantibodies and mononuclear cells and (2) recognition and elimination of cancerous cells. In contrast, harmful aspects include: (1) mounting a pathogenic autoimmune response against a tissue-derived ag, a 'modified self,' resulting in autoimmune disease and (2) inability to recognize and eliminate a cancerous clone. The immune system continuously faces internal and external influences; however, even when it is compromised or overwhelmed, it will still endeavor to regain and maintain tolerance to self. To promote this, we developed a 'modified vaccination technique' (MVT) (described as the third vaccination method after active and passive immunizations). It has two components: purified exogenous/endogenous ag (i.e., target ag) and a high-titer-specific antibody (ab) against the target ag made into an immune complex (IC) with predetermined immune-inducing components. The MVT works by ab information transfer (production of same class of immunoglobulin with the same specificity against the target ag that is present in the vaccine), thereby re-establishing tolerance to self (caused by exogenous/endogenous ags) following repeated administration of appropriate ICs. This vaccination technique can be used both prophylactically and therapeutically, and it mimics the immune system's natural abilities to respond to corrective information specifically, rapidly, safely and with minimal side effects and makes this approach a novel solution for many disorders that are difficult or impossible to cure or manage.
Collapse
|
40
|
Frascoli F, Flood E, Kim PS. A model of the effects of cancer cell motility and cellular adhesion properties on tumour-immune dynamics. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2017; 34:215-240. [PMID: 27094601 DOI: 10.1093/imammb/dqw004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/18/2016] [Indexed: 02/07/2023]
Abstract
We present a three-dimensional model simulating the dynamics of an anti-cancer T-cell response against a small, avascular, early-stage tumour. Interactions at the tumour site are accounted for using an agent-based model (ABM), while immune cell dynamics in the lymph node are modelled as a system of delay differential equations (DDEs). We combine these separate approaches into a two-compartment hybrid ABM-DDE system to capture the T-cell response against the tumour. In the ABM at the tumour site, movement of tumour cells is modelled using effective physical forces with a specific focus on cell-to-cell adhesion properties and varying levels of tumour cell motility, thus taking into account the ability of cancer cells to spread and form clusters. We consider the effectiveness of the immune response over a range of parameters pertaining to tumour cell motility, cell-to-cell adhesion strength and growth rate. We also investigate the dependence of outcomes on the distribution of tumour cells. Low tumour cell motility is generally a good indicator for successful tumour eradication before relapse, while high motility leads, almost invariably, to relapse and tumour escape. In general, the effect of cell-to-cell adhesion on prognosis is dependent on the level of tumour cell motility, with an often unpredictable cross influence between adhesion and motility, which can lead to counterintuitive effects. In terms of overall tumour shape and structure, the spatial distribution of cancer cells in clusters of various sizes has shown to be strongly related to the likelihood of extinction.
Collapse
Affiliation(s)
- Federico Frascoli
- Faculty of Science, Engineering and Technology, Department of Mathematics, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Emelie Flood
- School of Applied Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Peter S Kim
- School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| |
Collapse
|
41
|
Breast cancer vaccines delivered by dendritic cell-targeted lentivectors induce potent antitumor immune responses and protect mice from mammary tumor growth. Vaccine 2017; 35:5842-5849. [PMID: 28916248 DOI: 10.1016/j.vaccine.2017.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/15/2017] [Accepted: 09/04/2017] [Indexed: 12/13/2022]
Abstract
Breast cancer immunotherapy is a potent treatment option, with antibody therapies such as trastuzumab increasing 2-year survival rates by 50%. However, active immunotherapy through vaccination has generally been clinically ineffective. One potential means of improving vaccine therapy is by delivering breast cancer antigens to dendritic cells (DCs) for enhanced antigen presentation. To accomplish this in vivo, we pseudotyped lentiviral vector (LV) vaccines with a modified Sindbis Virus glycoprotein so that they could deliver genes encoding the breast cancer antigen alpha-lactalbumin (Lalba) or erb-b2 receptor tyrosine kinase 2 (ERBB2 or HER2) directly to resident DCs. We hypothesized that utilizing these DC-targeting lentiviral vectors asa breast cancer vaccine could lead to an improved immune response against self-antigens found in breast cancer tumors. Indeed, single injections of the vaccine vectors were able to amplify antigen-specific CD8T cells 4-6-fold over naïve mice, similar to the best published vaccine regimens. Immunization of these mice completely inhibited tumor growth in a foreign antigen environment (LV-ERBB2 in wildtype mice), and it reduced the rate of tumor growth in a self-antigen environment (LV-Lalba in wildtype or LV-ERBB2 in MMTV-huHER2 transgenic). These results show that a single injection with targeted lentiviral vectors can be an effective immunotherapy for breast cancer. Furthermore, they could be combined with other immunotherapeutic regimens to improve outcomes for patients with breast cancer.
Collapse
|
42
|
Zupančič E, Curato C, Paisana M, Rodrigues C, Porat Z, Viana AS, Afonso CAM, Pinto J, Gaspar R, Moreira JN, Satchi-Fainaro R, Jung S, Florindo HF. Rational design of nanoparticles towards targeting antigen-presenting cells and improved T cell priming. J Control Release 2017; 258:182-195. [PMID: 28511928 DOI: 10.1016/j.jconrel.2017.05.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 12/21/2022]
Abstract
Vaccination is a promising strategy to trigger and boost immune responses against cancer or infectious disease. We have designed, synthesized and characterized aliphatic-polyester (poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NP) to investigate how the nature of protein association (adsorbed versus entrapped) and polymer/surfactant concentrations impact on the generation and modulation of antigen-specific immune responses. The ability of the NP formulations to target dendritic cells (DC), be internalized and activate the T cells was characterized and optimized in vitro and in vivo using markers of DC activation and co-stimulatory molecules. Ovalbumin (OVA) was used as a model antigen in combination with the engraftment of CD4+ and CD8+ T cells, carrying a transgenic OVA-responding T cell receptor (TCR), to trace and characterize the activation of antigen-specific CD4+ and CD8+ lymph node T cells upon NP vaccination. Accordingly, the phenotype and frequency of immune cell stimulation induced by the NP loaded with OVA, isolated or in combination with synthetic unmethylated cytosine-phosphate-guanine (CpG) oligodeoxynucleotide (ODN) motifs, were characterized. DC-NP interactions increased with incubation time, presenting internalization values between 50 and 60% and 30-40%, in vitro and in vivo, respectively. Interestingly, animal immunization with antigen-adsorbed NP up-regulated major histocompatibility complex (MHC) class II (MHCII), while NP entrapping the antigen up-regulated MHCI, suggesting a more efficient cross-presentation. On the other hand, rather surprisingly, the surfactant used in the NP formulation had a major impact on the activation of antigen presenting cells (APC). In fact, DC collected from lymph nodes of animals immunized with NP prepared using poly(vinil alcohol) (PVA), as a surfactant, expressed significantly higher levels of CD86, MHCI and MHCII. In addition, those NP prepared with PVA and co-entrapping OVA and the toll-like receptor (TLR) ligand CpG, induced the most profound antigen-specific T cell response, by both CD4+ and CD8+ T cells, in vivo. Overall, our data reveal the impact of NP composition and surface properties on the type and extension of induced immune responses. Deeper understanding on the NP-immune cell crosstalk can guide the rational development of nano-immunotherapeutic systems with improved and specific therapeutic efficacy and avoiding off-target effects.
Collapse
Affiliation(s)
- Eva Zupančič
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.; CNC - Center for Neuroscience and Cell Biology University of Coimbra, Portugal; Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Caterina Curato
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Maria Paisana
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Catarina Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Ziv Porat
- Department of Biological Services, The Weizmann Institute of Science, Rehovot, Israel
| | - Ana S Viana
- Chemistry and Biochemistry Center, Sciences Faculty, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Carlos A M Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - João Pinto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Rogério Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - João N Moreira
- CNC - Center for Neuroscience and Cell Biology University of Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Israel
| | - Steffen Jung
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal..
| |
Collapse
|
43
|
Gupta J, Hoque M, Zaman M, Khan RH, Saleemuddin M. A detergent-based procedure for the preparation of IgG-like bispecific antibodies in high yield. Sci Rep 2016; 6:39198. [PMID: 27982091 PMCID: PMC5159798 DOI: 10.1038/srep39198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/14/2016] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies (BsAbs), with the ability to recognize two different epitopes simultaneously, offer remarkable advantages in bioassays, cancer therapy, biosensors, and enzyme electrodes. Preparation and purification of BsAbs in adequate quantities remains a major hurdle in their use in various applications. Poor yield is also the principal limitation in the preparation of BsAbs by the redox procedure. IgG with reduced inter-heavy chain disulfides do not dissociate into half molecules at neutral pH. In this study, we report that the dissociation occurs in presence of sodium dodecyl sulphate (SDS) and inclusion of the detergent during the redox procedure results in remarkable increase in the formation of the BsAbs. Exposure of antibodies to 0.1% (w/v) SDS causes only minor loss in secondary/tertiary structure and the ability to bind the antigen. The BsAbs prepared using the modified redox procedure that recognize the antigens HRP and α-LA were prepared and successfully employed for detecting α-LA in milk/dairy products by ELISA and dot blot techniques. BsAbs were also prepared from partially purified immunoglobulin gamma (IgG). This work shows for the first time that SDS, by dissociating IgG with reduced inter-heavy chain disulfides into half molecules, markedly enhances the formation of BsAbs by the redox procedure.
Collapse
Affiliation(s)
- Jyoti Gupta
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Mehboob Hoque
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Masihuz Zaman
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - M Saleemuddin
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
44
|
Bautz DJ, Sherpa AT, Threadgill DW. Prophylactic vaccination targeting ERBB3 decreases polyp burden in a mouse model of human colorectal cancer. Oncoimmunology 2016; 6:e1255395. [PMID: 28197371 PMCID: PMC5283616 DOI: 10.1080/2162402x.2016.1255395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 11/29/2022] Open
Abstract
Prophylactic vaccination is typically utilized for the prevention of communicable diseases such as measles and influenza but, with the exception of vaccines to prevent cervical cancer, is not widely used as a means of preventing or reducing the incidence of cancer. Here, we utilize a peptide-based immunotherapeutic approach targeting ERBB3, a pseudo-kinase member of the EGFR/ERBB family of receptor tyrosine kinases, as a means of preventing occurrence of colon polyps. Administration of the peptide resulted in a significant decrease in the development of intestinal polyps in C57BL/6J-ApcMin mice, a model of familial adenomatous polyposis (FAP). In addition, even though they were not vaccinated, ApcMin offspring born to vaccinated females developed significantly fewer polyps than offspring born to control females. Lastly, to validate ERBB as a valid target for vaccination, we found no overt toxicity, increases in apoptosis, or morphological changes in tissues where Erbb3 was ablated in adult mice. These results indicate that prophylactic vaccination targeting ERBB3 could prevent the development of colon polyps in an at-risk patient population.
Collapse
Affiliation(s)
- David J Bautz
- Department of Genetics, North Carolina State University , Raleigh, NC, USA
| | - Ang T Sherpa
- Department of Genetics, North Carolina State University , Raleigh, NC, USA
| | - David W Threadgill
- Department of Veterinary Pathobiology, Department of Molecular and Cellular Medicine, Texas A&M University , College Station, TX, USA
| |
Collapse
|
45
|
Campion CA, Soden D, Forde PF. Antitumour responses induced by a cell-based Reovirus vaccine in murine lung and melanoma models. BMC Cancer 2016; 16:462. [PMID: 27412241 PMCID: PMC4944504 DOI: 10.1186/s12885-016-2536-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The ever increasing knowledge in the areas of cell biology, the immune system and the mechanisms of cancer are allowing a new phase of immunotherapy to develop. The aim of cancer vaccination is to activate the host immune system and some success has been observed particularly in the use of the BCG vaccine for bladder cancer as an immunostimulant. Reovirus, an orphan virus, has proven itself as an oncolytic virus in vitro and in vivo. Over 80 % of tumour cell lines have been found to be susceptible to Reovirus infection and it is currently in phase III clinical trials. It has been shown to induce immune responses to tumours with very low toxicities. METHODS In this study, Reovirus was examined in two main approaches in vivo, in mice, using the melanoma B16F10 and Lewis Lung Carcinoma (LLC) models. Initially, mice were treated intratumourally (IT) with Reovirus and the immune responses determined by cytokine analysis. Mice were also vaccinated using a cell-based Reovirus vaccine and subsequently exposed to a tumourigenic dose of cells (B16F10 or LLC). Using the same cell-based Reovirus vaccine, established tumours were treated and subsequent immune responses and virus retrieval investigated. RESULTS Upregulation of several cytokines was observed following treatment and replication-competent virus was also retrieved from treated tumours. Varying levels of cytokine upregulation were observed and no replication-competent virus was retrieved in vaccine-treated mice. Prolongation of survival and delayed tumour growth were observed in all models and an immune response to Reovirus, either using Reovirus alone or a cell-based vaccine was also observed in all mice. CONCLUSION This study provides evidence of immune response to tumours using a cell-based Reovirus vaccine in both tumour models investigated, B16F10 and LLC, cytokine induction was observed with prolongation of survival in almost all cases which may suggest a new method for using Reovirus in the clinic.
Collapse
Affiliation(s)
- Ciorsdan A Campion
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Cork, Ireland. .,School of Microbiology, University College Cork, Cork, Ireland.
| | - Declan Soden
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Cork, Ireland
| | - Patrick F Forde
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
46
|
Targeted Vaccination against Human α-Lactalbumin for Immunotherapy and Primary Immunoprevention of Triple Negative Breast Cancer. Cancers (Basel) 2016; 8:cancers8060056. [PMID: 27322324 PMCID: PMC4931621 DOI: 10.3390/cancers8060056] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/25/2016] [Accepted: 06/06/2016] [Indexed: 01/06/2023] Open
Abstract
We have proposed that safe and effective protection against the development of adult onset cancers may be achieved by vaccination against tissue-specific self-proteins that are “retired” from expression at immunogenic levels in normal tissues as we age, but are overexpressed in emerging tumors. α-Lactalbumin is an example of a “retired” self-protein because its expression in normal tissues is confined exclusively to the breast during late pregnancy and lactation, but is also expressed in the vast majority of human triple negative breast cancers (TNBC)—the most aggressive and lethal form of breast cancer and the predominant form that occurs in women at high genetic risk including those with mutated BRCA1 genes. In anticipation of upcoming clinical trials, here we provide preclinical data indicating that α-lactalbumin has the potential as a vaccine target for inducing safe and effective primary immunoprevention as well as immunotherapy against TNBC.
Collapse
|
47
|
Wojtowicz ME, Dunn BK, Umar A. Immunologic approaches to cancer prevention-current status, challenges, and future perspectives. Semin Oncol 2015; 43:161-172. [PMID: 26970135 DOI: 10.1053/j.seminoncol.2015.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential of the immune system to recognize and reject tumors has been investigated for more than a century. However, only recently impressive breakthroughs in cancer immunotherapy have been seen with the use of checkpoint inhibitors. The experience with various immune-based strategies in the treatment of late cancer highlighted the importance of negative impact advanced disease has on immunity. Consequently, use of immune modulation for cancer prevention rather than therapy has gained considerable attention, with many promising results seen already in preclinical and early clinical studies. Although not without challenges, these results provide much excitement and optimism that successful cancer immunoprevention could be within our reach. In this review we will discuss the current state of predominantly primary and secondary cancer immunoprevention, relevant research, potential barriers, and future directions.
Collapse
Affiliation(s)
- Malgorzata E Wojtowicz
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Barbara K Dunn
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Kokate RA, Thamake SI, Chaudhary P, Mott B, Raut S, Vishwanatha JK, Jones HP. Enhancement of anti-tumor effect of particulate vaccine delivery system by 'bacteriomimetic' CpG functionalization of poly-lactic-co-glycolic acid nanoparticles. Nanomedicine (Lond) 2015; 10:915-29. [PMID: 25867857 DOI: 10.2217/nnm.14.144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM Low immunogenicity remains a major obstacle in realizing the full potential of cancer vaccines. In this study, we evaluated CpG-coated tumor antigen (Tag)-encapsulating 'bacteriomimetic' nanoparticles (CpG-nanoparticle [NP]-Tag NPs) as an approach to enhance anti-tumor immunity. MATERIALS & METHODS CpG-NP-Tag NPs were synthesized, characterized for their physicochemical properties and tested in vivo. RESULTS We found CpG predosing followed by intraperitoneal (IP) immunization with CpG-NP-Tag NPs significantly attenuated tumor growth in female BALB/c mice compared with respective controls. Histopathological and Immunofluorescence data revealed CpG-NP-Tag tumors had lower proliferation, higher apoptotic activity, greater CD4(+) and CD8(+) T cell infiltration as well as higher IFN-γ levels as compared with control groups. CONCLUSION Our findings suggest CpG-NP-Tag NPs can enhance anti-tumor effect of nanoparticulate tumor vaccination system.
Collapse
Affiliation(s)
- Rutika A Kokate
- Department of Molecular & Medical Genetics, University of North Texas Health Science Center (UNTHSC), Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Tuohy VK. Retired self-proteins as vaccine targets for primary immunoprevention of adult-onset cancers. Expert Rev Vaccines 2014; 13:1447-62. [DOI: 10.1586/14760584.2014.953063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
50
|
Chaudhuri A, Chattopadhyay A. Lipid binding specificity of bovine α-lactalbumin: A multidimensional approach. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2078-86. [DOI: 10.1016/j.bbamem.2014.04.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 10/25/2022]
|